EP2300446B1 - Production efficace de proteines heterologues a l'aide d'inhibiteurs de la mannosyl transferase - Google Patents

Production efficace de proteines heterologues a l'aide d'inhibiteurs de la mannosyl transferase Download PDF

Info

Publication number
EP2300446B1
EP2300446B1 EP09751270A EP09751270A EP2300446B1 EP 2300446 B1 EP2300446 B1 EP 2300446B1 EP 09751270 A EP09751270 A EP 09751270A EP 09751270 A EP09751270 A EP 09751270A EP 2300446 B1 EP2300446 B1 EP 2300446B1
Authority
EP
European Patent Office
Prior art keywords
protein
cell
linked glycosylation
culture
pmt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP09751270A
Other languages
German (de)
English (en)
Other versions
EP2300446A1 (fr
Inventor
Ranjit Desai
Lihu Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Sharp and Dohme LLC
Original Assignee
Merck Sharp and Dohme LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Sharp and Dohme LLC filed Critical Merck Sharp and Dohme LLC
Publication of EP2300446A1 publication Critical patent/EP2300446A1/fr
Application granted granted Critical
Publication of EP2300446B1 publication Critical patent/EP2300446B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione

Definitions

  • Glycoproteins mediate many essential functions in humans and other mammals, including catalysis, signaling, cell-cell communication, and molecular recognition and association. Glycoproteins make up the majority of non-cytosolic proteins in eukaryotic organisms ( Lis and Sharon, 1993, Eur. J. Biochem. 218:1-27 ). Many glycoproteins have been exploited for therapeutic purposes, and during the last two decades, recombinant versions of naturally-occurring glycoproteins have been a major part of the biotechnology industry.
  • recombinant glycosylated proteins used as therapeutics include erythropoietin (EPO), therapeutic monoclonal antibodies (mAbs), tissue plasminogen activator (tPA), interferon- ⁇ (IFN- ⁇ ), granulocyte-macrophage colony stimulating factor (GM-CSF), and human chorionic gonadotrophin (hCH) ( Cumming et al., 1991, Glycobiology 1:115-130 ). Variations in glycosylation patterns of recombinantly produced glycoproteins have recently been the topic of much attention in the scientific community as recombinant proteins produced as potential prophylactics and therapeutics approach the clinic.
  • EPO erythropoietin
  • mAbs therapeutic monoclonal antibodies
  • tPA tissue plasminogen activator
  • IFN- ⁇ interferon- ⁇
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • hCH human chorionic gonadotrophin
  • glycosylation structures of glycoprotein oligosaccharides will vary depending upon the host species of the cells used to produce them.
  • Therapeutic proteins produced in non-human host cells are likely to contain non-human glycosylation which may elicit an immunogenic response in humans-e.g. hypermannosylation in yeast ( Ballou, 1990, Methods Enzymol. 185:440-470 ); ⁇ (1,3)-fucose and ⁇ (1,2)-xylose in plants, ( Cabanes-Macheteau et al., 1999. Glycobiology, 9: 365-372 ); N-glycolylneuraminic acid in Chinese hamster ovary cells ( Noguchi et al., 1995. J. Biochem.
  • Carbohydrate chains bound to proteins in animal cells include N-glycoside bond type carbohydrate chains (also called N-glycans; or N-linked glycosylation) bound to an asparagine (Asn) residue in the protein and O-glycoside bond type carbohydrate chains (also called O-glycans; or O-linked glycosylation) bound to a serine (Ser) or threonine (Thr) residue in the protein.
  • glycoproteins produced by non-human mammalian cells tend to be more closely related to those of human glycoproteins
  • most commercial glycoproteins are produced in mammalian cells.
  • mammalian cells have several important disadvantages as host cells for protein production. Besides being costly, processes for producing proteins in mammalian cells produce heterogeneous populations of glycoforms, have low volumetric titers, and require both ongoing viral containment and significant time to generate stable cell lines.
  • glycosylation patterns of Igs are associated with different biological properties ( Jefferis and Lund, 1997, Antibody Eng. Chem. Immunol., 65: 111-128 ; Wright and Morrison, 1997, Trends Biotechnol., 15: 26-32 ).
  • glycoprotein compositions can vary with cell culture conditions, which may render some glycoprotein compositions immunogenic depending on the specific galactose pattern on the glycoprotein ( Patel et al., 1992. Biochem J. 285: 839-845 ).
  • specific glycoform(s) contribute(s) to a desired biological function
  • the ability to enrich for specific glycoforms on glycoproteins is highly desirable. Because different glycoforms are associated with different biological properties, the ability to enrich for glycoproteins having a specific glycoform can be used to elucidate the relationship between a specific glycoform and a specific biological function of the glycoprotein. Also, the ability to enrich for glycoproteins having a specific glycoform enables the production of therapeutic glycoproteins having particular specificities. Thus, production of glycoprotein compositions that are enriched for particular glycoforms is highly desirable.
  • O-glycosylation is a posttranslational event, which occurs in the cis-Golgi ( Varki, 1993, Glycobiol., 3: 97-130 ).
  • One gene family that has a role in O-linked glycosylation are the genes encoding the Dol-P-Man:Protein (Ser/Thr) Mannosyl Transferase (Pmt). These highly conserved genes have been identified in both higher eukaryotes such as humans, rodents, insects, and the like and lower eukaryotes such as fungi and the like. Yeast such as Saccharomyces cerevisiae and Pichia pastoris encode up to seven PMT genes encoding Pmt homologues (reviewed in Willer et al. Curr. Opin. Struct. Biol. 2003 Oct;13(5): 621-30 .).
  • O-linked glycosylation starts by the addition of the initial mannose from dolichol-phosphate mannose to a serine or threonine residue of a nascent glycoprotein in the endoplasmic reticulum by one of the seven O-mannosyl transferases genes. While there appear to be seven PMT genes encoding Pmt homologues in yeast, O-mannosylation of secreted fungal and heterologous proteins in yeast is primarily dependent on the genes encoding Pmt1 and Pmt2, which appear to function as a heterodimer. PMT1 and PMT2 and their protein products, Pmt1 and Pmt2, respectively, appear to be highly conserved among species.
  • Tanner et al. in U. S. Patent No. 5,714,377 describes the PMT1 and PMT2 genes of Saccharomyces cerevisiae and a method for making recombinant proteins having reduced O-linked glycosylation that uses fungal cells in which one or more of PMT genes have been genetically modified so that recombinant proteins are produced, which have reduced O-linked glycosylation.
  • Ng et al. in U.S. Published Patent Application No. 20020068325 discloses inhibition of O-glycosylation through the use of antisense or cosuppression or through the engineering of yeast host strains that have loss of function mutations in genes associated with O-linked glycosylation, in particular, one or more of the PMT genes.
  • UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetyl galactosaminyl-transferases (GalNAc-transferases) are involved in mucin type O-linked glycosylation found in higher eukaryotes. These enzymes initiate O-glycosylation of specific serine and threonine amino acids in proteins by adding N-acetylgalactosamine to the hydroxy group of these amino acids to which mannose residues can then be added in a step-wise manner. Clausen et al. in U. S. Patent No. 5,871,990 and U.S. Published Patent Application No.
  • 20050026266 discloses a family of nucleic acids encoding UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetyl galactosaminyl-transferases (GalNAc-transferases).
  • GaINAc-beta-benzyl discloses the use of GaINAc-beta-benzyl to selectively inhibit lectins of polypeptide GalNAc-transferases and not serve as substrates for other glycosyltransferases involved in O-glycan biosyntheses, thus inhibiting O-glycosylation.
  • Inhibitors of O-linked glycosylation have been described.
  • Orchard et al. in U.S. Patent No. 7,105,554 describes benzylidene thiazolidinediones and their use as antimycotic agents, e.g., antifungal agents.
  • These benzylidene thiazolidinediones are reported to inhibit the Pmt1 enzyme, preventing the formation of the O-linked mannoproteins and compromising the integrity of the fungal cell wall. The end result is cell swelling and ultimately death through rupture.
  • Konrad et al. in U.S. Published Patent Application No. 20020128235 disclose a method for treating or preventing diabetes mellitus by pharmacologically inhibiting O-linked protein glycosylation in a tissue or cell.
  • the method relies on treating a diabetic individual with (Z)-1-[N-(3-Ammoniopropyl)-N-(n-propyl)amino] diazen-ium-1,2-diolate or a derivative thereof, which binds O-linked N-acetylglucosamine transferase and thereby inhibits O-linked glycosylation.
  • Kojima et al. in U. S. Patent No. 5,268,364 disclose therapeutic compositions for inhibition of O-glycosylation using compounds such as benzyle- ⁇ -N-acetylgalactosamine, which inhibits extension of O-glycosylation leading to accumulation of O- ⁇ -GalNAc, to block expression of SLex or SLea by leukocytes or tumor cells and thereby inhibit adhesion of these cells to endothelial cells and platelets.
  • the invention is directed to novel inhibitors of Pmt proteins, which are useful for production of recombinant proteins with reduced O-linked glycosylation. This enables O-linked glycosylation of proteins produced from fungi and yeast cells to be controlled.
  • the method includes producing the protein in cells cultured in the presence of certain benzylidene thiazolidinediones inhibitors of Pmt-mediated O-linked glycosylation.
  • the present invention provides novel compounds and methods for expressing a recombinant protein (includes polypeptides and glycoproteins), which is susceptible to O-linked glycosylation in a particular host cell, having a reduced amount of O-linked glycosylation (including no O-linked glycosylation) in that cell type.
  • the method involves inducing expression of a protein of interest in a host cell in which the protein is susceptible to O-linked glycosylation in the host cell in the presence of one or more novel compounds of the invention which are inhibitors of the activity of one or more of the Dol-P-Man:Protein (Ser/Thr) Mannosyl Transferase (Pmt) proteins involved in the transfer of mannose to a serine or threonine residue of the protein in the cell, optionally with one or more ⁇ 1,2-mannosidases as described in Bobrowicz et al. U.S. Published Application No. 2007061631 , at the time expression of the protein is induced.
  • Dol-P-Man:Protein (Ser/Thr) Mannosyl Transferase (Pmt) proteins involved in the transfer of mannose to a serine or threonine residue of the protein in the cell, optionally with one or more ⁇ 1,2-mannosidases as described in Bobrowicz et
  • the protein that is expressed in the presence of the inhibitor has a reduced amount of O-linked glycosylation compared to the amount of O-linked glycosylation that would have been present on the protein if it had been produced in the absence of the inhibitor.
  • the method is particularly useful because it provides a means for producing therapeutically relevant proteins where it is desired that the protein have a reduced amount of O-glycosylation in host cells such as lower eukaryotes, for example yeast, and bacteria, which would normally produce proteins with O-linked glycans, having a reduced number of O-linked glycans.
  • the method is especially suitable for expressing proteins with reduced O-linked glycosylation in lower eukaryotic organisms, the method can also be practiced in higher eukaryotic organisms and bacteria.
  • the Pmt inhibitors of the invention are selected from the following group: and or a salt thereof.
  • Tanner et al. in U. S. Patent No. 5,714,377 describes a method for making recombinant proteins having reduced O-linked glycosylation using fungal cells such as yeast cells in which one or more of PMT genes encoding the Pmt protein have been genetically modified so that recombinant proteins are produced which have reduced O-linked glycosylation.
  • While deletion of the PMT1, PMT2, or PMT4 genes in a fungal host cell enables production of a recombinant protein having reduced O-linked glycosylation in the fungal host cell, expression of the PMT genes are important for host cell growth and either deletion alone also adversely affects the ability of the fungal host cell to grow thus making it difficult to produce a sufficient quantity of host cells or recombinant protein with a reduced amount of O-linked glycosylation.
  • Deletion of PMT2 plus either PMT1 or PMT4 appears to be lethal to the fungal host cell. Therefore, genetic elimination of the PMT genes in a host cell would appear to be an undesirable means for producing recombinant proteins having reduced O-linked glycosylation.
  • the PMT genes in the host cells used in the methods of the present invention have not been modified or deleted, which enables the host cell to O-glycosylate those proteins that are important for cell growth until which time the activity of the Pmt proteins is inhibited. In general, this enables the host cells to be grown to higher levels than the levels that could be obtained if the PMT genes had been deleted.
  • expression of the recombinant protein in the host cell is controlled by an inducible promoter and the Pmt activity in the host cell is not inhibited until expression of the recombinant protein is induced.
  • the Pmt inhibitors described herein are improved over those described previously in Orchard et al. (U.S. Patent No. 7,105,554 ) and Bobrowicz et al. (U.S. Published Application No. 2007061631 ) based on increased potency as shown in Example 5.
  • the increased potency allows for the use of smaller amounts of inhibitor to reduce fungal O-glycosylation to acceptable levels and/or increases the potential for complete elimination of O-glycans.
  • the methods described herein facilitate the production of glycoproteins having reduced O-linked glycosylation in host cells that have been genetically modified to produce glycoproteins having predominantly a particular N-linked glycan structure but which also O-glycosylate the glycoprotein.
  • Methods for producing a wide variety of glycoproteins having predominantly particular N-linked glycoforms have been disclosed in U.S. Patent No. 7,029,872 and U.S. Published Application Nos. 20050170452 , 20050260729 , 20040230042 , 20050208617 , 20050208617 , 20040171826 , 20060160179 , 20060040353 , and 20060211085 .
  • Any one of the host cells described in the aforementioned patent and patent applications can be used to produce a glycoprotein having predominantly a particular N-linked glycan structure and having reduced O-linked glycosylation using the method disclosed herein. It has been found that some host cells that have been genetically modified to produce glycoproteins having predominantly a particular N-linked glycan structure can grow less well in culture under particular conditions than host cells that have not been modified. For example, particular fungal and yeast cells in which genes involved in hypermannosylation have been deleted and other genes needed to produce particular mammalian or human like N-linked glycan structures have been added, can grow less well than fungal or yeast cells that do not the genetic modifications.
  • deletions of the PMT genes either is lethal to the cells or adversely affects the ability of the cells to grow to sufficient quantities in culture.
  • the methods herein avoid the potential deleterious effects of deleting the PMT genes by allowing the cells to grow to sufficient quantities in culture before inducing expression of the recombinant glycoprotein and adding an inhibitor of the activity of the Pmt proteins, optionally with one or more ⁇ 1,2-mannosidases, to produce the recombinant glycoprotein having predominantly particular N-linked glycan structures and reduced O-linked glycosylation.
  • An aspect of the methods described herein is that it provides for a glycoprotein composition comprising reduced O-linked glycosylation and a predominantly a specific N-linked glycoform in which the recombinant glycoprotein may exhibit increased biological activity and/or decreased undesired immunogenicity relative to compositions of the same glycoprotein produced from mammalian cell culture, such as CHO cells.
  • An additional advantage of producing the glycoprotein composition comprising reduced O-linked glycosylation and a predominant N-linked glycoform is that it avoids production of undesired or inactive glycoforms and heterogeneous mixtures, which may induce undesired effects and/or dilute the more effective glycoform.
  • therapeutic pharmaceutical composition of glycoprotein molecules comprising, for example, predominantly Man 5 GlcNAc 2 , Man 3 GlcNAc 2 , GlcNAcMan 5 GlcNAc 2 , GlcNAcMan 3 GlcNAC 2 , GlcNAc 2 Man 3 GlcNAc 2 , GalGlcNAcMan 5 GlcNAc 2 , Gal(GlcNAc) 2 Man 5 GlcNAc 2 , (GalGlcNAc) 2 Man 5 GlcNAc 2 , NANAGalGlcNAcMan 3 GlcNAc 2 , NANA 2 Gal 2 GlcNAcMan 3 GlcNAc 2 , and GalGlcNAcMan 3 GlcNAc 2 glycoforms and having reduced O-linked glycosylation may well be effective at lower doses, thus having higher efficacy/potency.
  • the method for producing proteins having reduced O-linked glycosylation comprises transforming a host cell with a nucleic acid encoding a recombinant or heterologous protein in which it is desirable to produce the protein having reduced O-linked glycosylation.
  • the nucleic acid encoding the recombinant protein is operably linked to regulatory sequences that allow expression of the recombinant protein.
  • regulatory sequences include an inducible promoter and optionally an enhancer upstream, or 5', to the nucleic acid encoding the fusion protein and a transcription termination site 3' or down stream from the nucleic acid encoding the recombinant protein.
  • the nucleic acid also typically encodes a 5' UTR region having a ribosome binding site and a 3' untranslated region.
  • the nucleic acid is often a component of a vector replicable in cells in which the recombinant protein is expressed.
  • the vector can also contain a marker to allow recognition of transformed cells.
  • some cell types, particularly yeast can be successfully transformed with a nucleic acid lacking extraneous vector sequences.
  • Nucleic acids encoding desired recombinant proteins can be obtained from several sources. cDNA sequences can be amplified from cell lines known to express the protein using primers to conserved regions (see, for example, Marks et al., J. Mol. Biol. 581-596 (1991 )). Nucleic acids can also be synthesized de novo based on sequences in the scientific literature. Nucleic acids can also be synthesized by extension of overlapping oligonucleotides spanning a desired sequence (see, e.g., Caldas et al., Protein Engineering, 13, 353-360 (2000 )).
  • the nucleic acid encoding the protein is operably linked to an inducible promoter, which allows expression of the protein to be induced when desired.
  • the nucleic acid encoding the protein is operably linked to a constitutive promoter.
  • the protein include a signal sequence that directs the protein to be excreted into the cell culture medium where it can then be isolated.
  • the transformed host cells are cultured for a time sufficient to produce a desired multiplicity of host cells sufficient to produce the desired amount of protein before adding one or more inhibitors of Pmt-mediated O-linked glycosylation to the culture medium.
  • the inducer and inhibitor can be added to the culture simultaneously or the inducer is added to the culture before adding the one or more Pmt inhibitors or the one or more Pmt inhibitors is added to the culture before adding the inducer.
  • the induced protein is produced having reduced O-linked glycosylation and can be recovered from the culture medium or for proteins not having a signal sequence, from the host cell by lysis.
  • the one or more inhibitors of Pmt-mediated O-linked glycosylation is added to the culture medium at the same time the culture is established and the protein, which is produced having reduced O-linked glycosylation, can be recovered from the culture medium or for proteins not having a signal sequence, from the host cell by lysis.
  • the inhibitor inhibits at least the activity of Pmt1 or Pmt2, or both.
  • the inhibitor inhibits activity of the homologue in the higher eukaryote that corresponds to the Pmt1 or Pmt2.
  • the compounds of the invention are shown to be effective in producing recombinant proteins having reduced O-linked glycosylation in Pichia pastoris strains that had intact, functional PMT genes.
  • Table 1 and Figure 1 of Example 5 show that any one of the above four Pmt chemical inhibitors added to a culture of recombinant Pichia pastoris having intact, functional PMT genes and transformed with a nucleic acid encoding a recombinant, human anti-Her2 antibody protein operably linked to an inducible promoter at the time of expression of the recombinant protein was induced, produced a recombinant protein having a level of reduced O-linked glycosylation that was improved relative to the level of O-linked glycosylation seen for Pichia pastoris cells treated with Pmt inhibitor Pmti-3 described in Orchard et al. (Bioorgan & Med Chem Letters (2004) 14:3975-3978 ) and patent publications by the same authors including EP 1313471 B1 and Bobrowicz et al.
  • cells refers to host cells described as follows. While host cells for the method herein includes both higher eukaryote cells and lower eukaryote cells, lower eukaryote cells, for example filamentous fungi or yeast cells, are currently preferred for expression of proteins because they can be economically cultured, give high yields of protein, and when appropriately modified are capable of producing proteins having suitable glycosylation patterns.
  • Lower eukaryotes include yeast, fungi, collar-flagellates, microsporidia, alveolates (e.g., dinoflagellates), stramenopiles (e.g, brown algae, protozoa), rhodophyta (e.g., red algae), plants (e.g., green algae, plant cells, moss) and other protists.
  • Yeast and fungi include, but are not limited to: Pichia sp.
  • Pichia pastoris for example, Pichia pastoris , Pichia finlandica , Pichia trehalophila , Pichia koclamae, Pichia membranaefaciens, Pichia minuta ( Ogataea minuta, Pichia lindneri), Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica ), Saccharomyces sp. (for example Saccharomyces cerevisiea ), Hansenula polymorpha , Kluyveromyces sp.
  • yeast in particular, are currently preferred because yeast offers established genetics allowing for rapid transformations, tested protein localization strategies, and facile gene knock-out techniques.
  • Suitable vectors have expression control sequences, such as promoters, including 3-phosphoglycerate kinase or other glycolytic enzymes, and an origin of replication, termination sequences, and the like as desired.
  • yeasts such as K. lactis , Pichia pastoris , Pichia methanolica , and Hansenula polymorpha are currently preferred for cell culture because they are able to grow to high cell densities and secrete large quantities of recombinant protein.
  • filamentous fungi such as Aspergillus niger , Fusarium sp, Neurospora crass , and others can be used to produce recombinant proteins at an industrial scale.
  • Lower eukaryotes in particular filamentous fungi and yeast, can be genetically modified so that they express proteins or glycoproteins in which the glycosylation pattern is human-like or humanized. This can be achieved by eliminating selected endogenous glycosylation enzymes and/or supplying exogenous enzymes as described by Gerngross et al. in U.S. Patent No. US7029872 , and U. S. Published Patent Application Nos. 20040018590 , 20050170452 , 20050260729 , 20040230042 , 20050208617 , 20040171826 , 20050208617 , 20060160179 , 20060040353 , and 20060211085 .
  • a host cell can additionally or alternatively be engineered to express one or more enzymes or enzyme activities, which enable the production of particular N-glycan structures at a high yield.
  • Such an enzyme can be targeted to a host subcellular organelle in which the enzyme will have optimal activity, for example, by means of signal peptide not normally associated with the enzyme.
  • Host cells can also be modified to express a sugar nucleotide transporter and/or a nucleotide diphosphatase enzyme. The transporter and diphosphatase improve the efficiency of engineered glycosylation steps, by providing the appropriate substrates for the glycosylation enzymes in the appropriate compartments, reducing competitive product inhibition, and promoting the removal of nucleoside diphosphates. See, for example, Gerngross et al. in U.S. Published Patent Application No.20040018590 and Hamilton, 2003, Science 301: 1244-46 and the aforementioned U.S. patent and patent applications.
  • a host cell for example, yeast or fungal
  • yeast or fungal can be selected or engineered to be depleted in 1,6-mannosyl transferase activities, which would otherwise add mannose residues onto the N-glycan of a glycoprotein, and to further include a nucleic acid for ectopic expression of an ⁇ -1,2 mannosidase activity, which enables production of recombinant glycoproteins having greater than 30 mole percent Man 5 GlcNAC 2 N-glycans.
  • the host cells When a glycoprotein is produced in the host cells according to the method described herein, the host cells will produce a glycoprotein having predominantly a Man 5 GlcNAc 2 N-glycan structure and reduced O-glycosylation compared to the glycoprotein produced in the cell otherwise.
  • the host cell is engineered to further include a nucleic acid for ectopic expression of GlcNAc transferase I activity, which enables production of glycoproteins having predominantly GlcNAcMan5GlcNAc2 N-glycans.
  • the host cells When a glycoprotein is produced in the host cells according to the method described herein, the host cells will produce a glycoprotein having predominantly a GlcNAcMan 5 GlcNAc 2 N-glycan structure and reduced O-glycosylation compared to the glycoprotein produced in the cell otherwise.
  • the host cell is engineered to further include a nucleic acid for ectopic expression of mannosidase II activity, which enables production of glycoproteins having predominantly GlcNAcMan 3 GlcNAc 2 N-glycans.
  • the host cells When a glycoprotein is produced in the host cells according to the method described herein, the host cells will produce a glycoprotein having predominantly a GlcNAcMan 3 GlcNAc 2 N-glycan structure and reduced O-glycosylation compared to the glycoprotein produced in the cell otherwise.
  • the host cell is engineered to further include a nucleic acid for ectopic expression of GlcNAc transferase II activity, which enables production of glycoproteins having predominantly GlcNAc 2 Man 3 GlcNAc 2 N-glycans.
  • the host cells When a glycoprotein is produced in the host cells according to the method described herein, the host cells will produce a glycoprotein having predominantly a GlcNAc 2 Man 3 GlcNAc 2 N-glycan structure and reduced O-glycosylation compared to the glycoprotein produced in the cell otherwise.
  • the above host cells can be further engineered to produce particular hybrid or complex N-glycan or human-like N-glycan structures by further including one or more higher eukaryote genes involved in N-linked glycosylation, in any combination, that encode for example, sialytransferase activities, class II and III mannosidase activities, GlcNAc transferase II, III, IV, V, VI, IX activity, and galactose transferase activity. It is currently preferable that the cells further include one or more of nucleic acids encoding UDP-specific diphosphatase activity, GDP-specific diphosphatase activity, and UDP-GlcNAc transporter activity.
  • Plants and plant cell cultures may be used for expression of proteins and glycoproteins with reduced O-linked glycosylation as taught herein (See, for example, Larrick & Fry, 1991, Hum. Antibodies Hybridomas 2: 172-89 ); Benvenuto et al., 1991, Plant Mol. Biol. 17: 865-74 ); Durin et al., 1990, Plant Mol. Biol. 15: 281-93 ); Hiatt et al., 1989, Nature 342: 76-8 ).
  • Preferable plant hosts include, for example, Arabidopsis , Nicotiana tabacum , Nicotiana rustica , and Solanum tuberosum .
  • Insect cell culture can also be used to produce proteins and glycoproteins proteins and glycoproteins with reduced O-linked glycosylation, as taught herein for example, baculovirus-based expression systems (See, for example, Putlitz et al., 1990, Bio/Technology 8: 651-654 ).
  • mammalian tissue cell culture can also be used to express and produce proteins and glycoproteins with reduced O-linked glycosylation as taught herein ( See Winnacker, From Genes to Clones (VCH Publishers, NY, 1987 ).
  • Suitable hosts include CHO cell lines, various COS cell lines, HeLa cells, preferably myeloma cell lines or the like or transformed B-cells or hybridomas.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer ( Queen et al., 1986I, mmunol. Rev.
  • Expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, bovine Papilloma Virus, cytomegalovirus and the like.
  • a selectable marker such as a neoR expression cassette, is included in the expression vector.
  • the nucleic acid encoding the protein to be expressed can be transferred into the host cell by conventional methods, which vary depending on the type of cellular host. For example, calcium phosphate treatment, protoplast fusion, natural breeding, lipofection, biolistics, viral-based transduction, or electroporation can be used for cellular hosts. Tungsten particle ballistic transgenesis is preferred for plant cells and tissues. (See, generally, Maniatis et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Press, 1982 ))
  • the proteins or glycoproteins having reduced O-linked glycosylation can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like (See, generally, Scopes, R., Protein Purification (Springer-Verlag, N.Y., 1982 )). Substantially pure glycoproteins of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity most preferred, for pharmaceutical uses. Once purified, partially or to homogeneity as desired, the proteins can then be used therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings, and the like. ( See , generally, Immunological Methods, Vols. I and II (Lefkovits and Pernis, eds., Academic Press, NY, 1979 and 1981 ).
  • glycoprotein compositions comprising a predominant species of N-glycan structure and having reduced O-linked glycosylation compared to compositions of the glycoprotein which have been produced in host cells have not been incubated in the presence of an inhibitor of Pmt-mediated O-linked glycosylation or an ⁇ -1,2-mannosidase capable of trimming more than one mannose residue from a glycans structure or both.
  • the glycoprotein composition comprises a glycoprotein having a predominant N-glycan structure selected from the group consisting of Man 5 GlcNAc 2, Man 3 GlcNAc 2 , GlcNAcMan 5 GlcNAc 2 , GlcNAcMan 3 GlcNAc 2 , GlcNAc 2 Man 3 GlcNAc 2 , GalGlcNAcMan 5 GlcNAc 2 , Gal(GlcNAc) 2 Man 5 GlcNAc 2 , (GalGlcNAc) 2 Man 5 GlcNAc 2 , NANAGalGlcNAcMan 3 GlcNAc 2 , NANA 2 Gal 2 GlcNAcMan 3 GlcNAc 2 , and GalGlcNAcMan 3 GlcNAc 2 glycoforms.
  • a glycoprotein having a predominant N-glycan structure selected from the group consisting of Man 5 GlcNAc 2, Man 3 GlcNAc 2 , GlcNAcMan 5 Glc
  • compositions comprising the glycoprotein as an active therapeutic agent and a variety of other pharmaceutically acceptable components ( See , Remington's Pharmaceutical Science (15th ed., Mack Publishing Company, Easton, Pennsylvania, 1980 ). The preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination.
  • compositions or formulation can also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers, and the like.
  • compositions for parenteral administration are sterile, substantially isotonic, pyrogen-free and prepared in accordance with GMP of the FDA or similar body.
  • Glycoproteins can be administered as injectable dosages of a solution or suspension of the substance in a physiologically acceptable diluent with a pharmaceutical carrier that can be a sterile liquid such as water oils, saline, glycerol, or ethanol.
  • a pharmaceutical carrier can be a sterile liquid such as water oils, saline, glycerol, or ethanol.
  • auxiliary substances such as wetting or emulsifying agents, surfactants, pH buffering substances and the like can be present in compositions.
  • Other components of pharmaceutical compositions are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, and mineral oil.
  • glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.
  • Glycoproteins can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained release of the active ingredient.
  • compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above ( See Langer, Science 249, 1527 (1990 ) and Hanes, Advanced Drug Delivery Reviews 28, 97-119 (1997 ).
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates.
  • Salts derived from bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins such as arginine, be
  • the compounds of the present invention may contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers.
  • the present invention is meant to comprehend all such isomeric forms of these compounds.
  • EXAMPLE 4 The preparation of EXAMPLE 4 is the same as that for EXAMPLE 3 .
  • 1 H-NMR 400 MHz, MeOD
  • ⁇ 7.49 (s, 1H), 7.24-7.40 (m, 7H), 6.98-7.05 (m, 4H), 6.90-6.91 (m, 1H), 5.37-5.40 (m, 1H), 4.75 (s, 2H), 4.21-4.32 (m, 2H), 3.83-3.88 (m, 1H), 3.69-3.72 (m, 1H), 3.33-3.35 (m, 2H), 3.09-3.13 (t, J 6.57 Hz, 2H), 1.79-1.87 (m, 1H), 0.86-0.88(m, 6H).
  • Expression/integration plasmid vector pGLY2988 contains expression cassettes under control of the methanol-inducible Pichia pastoris AOX1 promoter that encode the heavy (Hc) and light (Lc) chains of anti-Her2.
  • Anti-Her2 Hc and Lc fused at the N-terminus to ⁇ -MAT pre signal peptide (SEQ ID Nos:1 and 2) were synthesized by GeneArt AG. Each was synthesized with unique 5' Eco R1 and 3' Fse 1 sites. The nucleotide and amino acid sequences of the anti-Her2 Hc are shown in SEQ ID Nos:3 and 4, respectively.
  • nucleotide and amino acid sequences of the anti-Her2 Lc are shown in SEQ ID Nos:5 and 6, respectively.
  • Both nucleic acid fragments encoding the Hc and Lc proteins fused to the ⁇ -MAT pre signal peptide were separately subcloned using 5' Eco R1 and 3' Fse 1 unique sites into an expression plasmid vector pGLY2198, which contains the Pichia pastoris TRP2 targeting nucleic acid and the Zeocin-resistance marker and generates expression cassettes under the control of the AOX1 promoter and Saccharomyces cerevisiae CYC terminator, to form plasmid vectors pGLY2987 and pGLY2338, respectively.
  • the Lc expression cassette was then removed from plasmid vector pGLY2338 by digesting with Bam HI and Not I and subcloned into plasmid vector pGLY2987 digested with Bam HI and Not 1, thus generating the final expression plasmid vector pGLY2988.
  • Anti-Her2 expression strain yGLY4280 was constructed as follows: Five micrograms of pGLY2988 digested with restriction enzyme Spe 1 which cuts in the TRP 2 targeting region were used to transform strain yGLY22-1. Strain yGLY22-1 ( och1 ⁇ :: lacZ bmt2 ⁇ ::lacZ / KlMNN2-2 / mnn4L1 ⁇ .:: / acZ / MmSLC35A3 pnol ⁇ mnn4 ⁇ ::lacZ met16 ⁇ :: / acZ ), was constructed using methods described earlier ( Nett and Gerngross, Yeast 20:1279 (2003 ); Choi et al., PNAS USA 100:5022 (2003 ); Hamilton et al., Science 301:1244 (2003 )).
  • Transformation of yGLY22-1 performed essentially as follows: YGLY22-1 was grown in 50 mL YPD media (yeast extract (1%), peptone (2%), dextrose (2%)) overnight to an OD of between about 0.2 to 6. After incubation on ice for 30 minutes, cells were pelleted by centrifugation at 2500-3000 rpm for 5minutes. Media was removed and the cells washed three times with ice cold sterile 1M sorbitol before resuspension in 0.5 ml ice cold sterile 1M sorbitol. Ten ⁇ L of linearized DNA (10 ug) and 100 ⁇ L cell suspension was combined in an electroporation cuvette and incubated for 5 minutes on ice.
  • Electroporation was in a Bio-Rad GenePulser Xcell following the preset Pichia pastoris protocol (2 kV, 25 ⁇ F, 200 ⁇ ), immediately followed by the addition of 1 mL YPDS recovery media (YPD media plus 1 M sorbitol). The transformed cells were allowed to recover for four hours to overnight at room temperature (26°C) before plating the cells on the selective media. Following selection on media containing zeocin, transformants were screened by small scale expression analysis to detect anti-Her2 expression. Strain yGLY4280 was selected based on high level anti-Her2 expression.
  • Anti-Her2 protein expression for strain yGLY4280 was carried out in shake flasks at 24°C with buffered glycerol-complex medium (BMGY) consisting of 1% yeast extract, 2% peptone, 100 mM potassium phosphate buffer pH 6.0, 1.34% yeast nitrogen base, 4 x 10-5 % biotin, and 1% glycerol.
  • BMGY buffered glycerol-complex medium
  • the induction medium for protein expression was buffered methanol-complex medium (BMMY) consisting of 1% methanol instead of glycerol in BMGY.
  • Pmt inhibitors in 100% methanol were added to the growth medium to a final concentration of 0.15 ug/mL at the time the induction medium was added.
  • O-glycan determination was performed using a Dionex-HPLC (HPAEC-PAD) as follows.
  • HPAEC-PAD Dionex-HPLC
  • protein was purified from the growth medium using protein A chromatography ( Li et al. Nat. Biotechnol. 24(2):210-5 (2006 )) and the O-glycans released from and separated from protein by alkaline elimination (beta -elimination) ( Harvey, Mass Spectrometry Reviews 18: 349- 451 (1999 )).
  • This process also reduces the newly formed reducing terminus of the released O-glycan (either oligomannose or mannose) to mannitol.
  • the mannitol group thus serves as a unique indicator of each O- glycan.
  • the sample was treated with 25 ⁇ L alkaline borohydride reagent and incubated at 50°C for 16 hours. About 20 uL arabitol internal standard was added, followed by 10 ⁇ L glacial acetic acid. The sample was then centrifuged through a Millipore filter containing both SEPABEADS and AG 50W-X8 resin and washed with water. The samples, including wash, were transferred to plastic autosampler vials and evaporated to dryness in a centrifugal evaporator.
  • Figure 1 Visual proof that the novel Pmt inhibitors effectively reduce O-glycosylation of Pichia -produced recombinant protein is provided by Figure 1 , which shows the effects of increasing amounts of Pmt inhibitors on O-glycosylation of anti-Her2 heavy chain (Hc).
  • Strain yGLY4280 was inoculated into 96-well deep well plates (Qiagen, Valencia, CA) containing 0.5ml of BMGY media per well. After 24 hours growth with vigorous shaking, the 96-well plate was centrifuged at 2,000 rpm for five minutes to pellet cells.
  • the media was removed and, following a wash step with 0.5mL of BMMY media, the cells resuspended in 0.2 mL BMMY media in which Pmt inhibitors were diluted 2-fold across the rows (11 wells).
  • Well #1 contained 5ug/mL of inhibitor
  • well # 2 contained 2.5 ug/mL and so on until well # 10 contained 0.009 ug/mL; well #11 contained no inhibitor.
  • the plate was centrifuged at 2,000 rpm for five minutes to pellet cells, and the cleared supernatant subjected to Western blot analysis to detect anti-Her2 expression.
  • the Western blotting was performed as follows: seven ⁇ L of the supernatants were separated by reducing polyacrylamide gel electrophoresis (SDS-PAGE) according to Laemmli, U. K. (1970) Nature 227, 680-685 and then electroblotted onto nitrocellulose membranes (Schleicher & Schuell, now Whatman, Inc., Florham Park, NJ). Anti-Her2 antibody chains were detected on the Western blots using a peroxidase-conjugated anti-human Hc and Lc antibody (Calbiochem/EMD Biosciences, La Jolla, CA) and developed using the ImmunoPure Metal Enhanced DAB Substrate Kit (Pierce Biotechnology, Rockford, IL).
  • SDS-PAGE polyacrylamide gel electrophoresis
  • Figure 1 shows the results of one such analysis in which the novel Pmt inhibitor from Example 4 (EX. 4, panel A) was tested against Pmti-3 from Orchard et al. (panel B) and also an inactive compound as a control (panel C).
  • Example 4 and Pmti-3 effectively reduced O-glycosylation of anti-Her2 Hc at concentrations as low as 0.018 ug/mL.
  • the inactive control compound panel C
  • Similar results were obtained with inhibitors shown in Examples 1, 2, and 3. Taken together, these results indicate that the novel Pmt inhibitors shown in Examples 1 to 4 are effective inhibitors of fungal O-glycosylation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Enzymes And Modification Thereof (AREA)

Claims (15)

  1. Composé sélectionné parmi le groupe suivant:
    Figure imgb0038
    Figure imgb0039
    Figure imgb0040
    Figure imgb0041
    ou un sel de celui-ci.
  2. Procédé de production d'une protéine ayant une glycosylation à liaison O réduite, comprenant:
    (a) cultiver une cellule qui produit la protéine en culture;
    (b) mettre la culture en contact avec un ou plusieurs composés selon la revendication 1, ce qui inhibe la glycosylation à liaison O médiée par Pmt; et
    (c) isoler la protéine produite par la cellule hôte.
  3. Procédé selon la revendication 2, dans lequel la culture de la cellule est assurée en:
    (a) fournissant un acide nucléique codant une protéine; et
    (b) introduisant l'acide nucléique dans la cellule.
  4. Procédé selon la revendication 3, dans lequel la culture est cultivée pendant suffisamment longtemps pour donner une multiplicité de cellules ayant l'acide nucléique avant de mettre la culture en contact avec l'un quelconque de l'un ou des plusieurs composés qui inhibent la glycosylation à liaison O médiée par Pmt.
  5. Procédé selon la revendication 3, dans lequel la culture est cultivée en présence de l'un quelconque de l'un ou des plusieurs composés qui inhibent la glycosylation à liaison O médiée par Pmt.
  6. Procédé selon la revendication 3, dans lequel l'acide nucléique est lié d'une manière opérationnelle à un promoteur inductible.
  7. Procédé selon la revendication 6, dans lequel la culture est cultivée pendant suffisamment longtemps pour donner une multiplicité de cellules ayant l'acide nucléique avant de mettre la culture en contact avec l'un ou les plusieurs composés qui inhibent la glycosylation à liaison O médiée par Pmt et un inducteur du promoteur afin d'induire l'expression de la protéine et d'isoler la protéine produite par la cellule en présence de l'un ou des plusieurs inhibiteurs et de l'inducteur pour produire la protéine ayant une glycosylation à liaison O réduite.
  8. Procédé selon la revendication 6, dans lequel la culture est mise en contact avec un inducteur du promoteur pour induire l'expression de la protéine pendant un certain temps avant de mettre la culture en contact avec l'un ou les plusieurs composés qui inhibent la glycosylation à liaison O médiée par Pmt et d'isoler la protéine produite par la cellule en présence de l'un ou des plusieurs composés qui inhibent la glycosylation à liaison O médiée par Pmt et de l'inducteur pour produire la protéine ayant une glycosylation à liaison O réduite.
  9. Procédé selon la revendication 3, dans lequel la cellule est une cellule fongique.
  10. Procédé selon la revendication 3, dans lequel la cellule est une cellule de levure.
  11. Procédé selon la revendication 3, dans lequel la cellule est sélectionnée parmi le groupe consistant en K. lactis, Pichia pastoris, Pichia methanolica et Hansenula.
  12. Procédé selon la revendication 3, dans lequel la cellule est Pichia pastoris.
  13. Procédé selon la revendication 3, dans lequel la cellule est une cellule de levure ou une cellule fongique filamenteuse qui a été génétiquement modifiée pour produire des glycoprotéines dotées d'une glycoforme N-glycane prédominante.
  14. Procédé selon la revendication 3, dans lequel les cellules ont été génétiquement modifiées pour produire des glycoprotéines dans lesquelles le modèle de N-glycosylation est pseudo-humain ou humanisé.
  15. Procédé selon la revendication 3, dans lequel la protéine est produite à un rendement d'au moins 100 mg/litre de milieu de culture.
EP09751270A 2008-05-20 2009-05-18 Production efficace de proteines heterologues a l'aide d'inhibiteurs de la mannosyl transferase Active EP2300446B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12825108P 2008-05-20 2008-05-20
PCT/US2009/044297 WO2009143041A1 (fr) 2008-05-20 2009-05-18 Production efficace de protéines hétérologues à l'aide d'inhibiteurs de la mannosyl transférase

Publications (2)

Publication Number Publication Date
EP2300446A1 EP2300446A1 (fr) 2011-03-30
EP2300446B1 true EP2300446B1 (fr) 2011-12-07

Family

ID=40929664

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09751270A Active EP2300446B1 (fr) 2008-05-20 2009-05-18 Production efficace de proteines heterologues a l'aide d'inhibiteurs de la mannosyl transferase

Country Status (9)

Country Link
US (2) US8309325B2 (fr)
EP (1) EP2300446B1 (fr)
JP (2) JP5411929B2 (fr)
CN (1) CN102036974B (fr)
AT (1) ATE536347T1 (fr)
AU (1) AU2009249314A1 (fr)
CA (1) CA2721459A1 (fr)
MX (1) MX2010012626A (fr)
WO (1) WO2009143041A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2628725A1 (fr) * 2005-11-15 2007-05-31 Glycofi, Inc. Production de glycoproteines a o-glycosylation reduite
MX2010012626A (es) * 2008-05-20 2011-02-22 Merck Sharp & Dohme Produccion eficiente de proteinas heterologas utilizando inhibidores de manosil transferasa.
CN103764837A (zh) 2011-02-25 2014-04-30 默沙东公司 用于生产具有修饰的o-糖基化的蛋白的酵母菌株
KR102111078B1 (ko) 2013-07-04 2020-05-18 노파르티스 아게 O-만노실트랜스퍼라제 결핍 사상균 세포 및 이의 이용 방법
SG11201700446XA (en) 2014-07-21 2017-02-27 Glykos Finland Oy Production of glycoproteins with mammalian-like n-glycans in filamentous fungi

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4318550A1 (de) 1993-06-04 1994-12-08 Boehringer Mannheim Gmbh Aryliden-4-oxo-2-thioxo-3- thiazolidincarbonsäuren, Verfahren zu ihrer Herstellung und diese Verbindungen enthaltende Arzneimittel
US6103501A (en) * 1997-11-17 2000-08-15 Washington University Single chain glycoprotein hormones comprising two β and one α subunits and recombinant production thereof
FR2804113B1 (fr) * 2000-01-26 2004-06-18 Lipha Derives animes de dihydro-1,3,5-triazine et leurs applications en therapeutique
US6506755B2 (en) * 2000-02-03 2003-01-14 Hoffmann-La Roche Inc. Thiazolidinecarboxyl acids
US20030186948A1 (en) * 2000-03-21 2003-10-02 Jeffrey Kudlow O-linked N-acetylglucosamine pathway in the pathogenesis of neurodegeneration and diabetes
WO2001070236A1 (fr) * 2000-03-21 2001-09-27 Uab Research Foundation Prevention et/ou traitement du diabete sucre par inhibition pharmacologique de la glycosylation de la proteine a liaison o de cellule beta pancreatique et/ou de la o-glycosylation p135 de cellule beta pancreatique
GB0021419D0 (en) * 2000-08-31 2000-10-18 Oxford Glycosciences Uk Ltd Compounds
GB0021421D0 (en) * 2000-08-31 2000-10-18 Oxford Glycosciences Uk Ltd Compounds
EP1456187A4 (fr) * 2001-11-15 2005-02-09 Incyte San Diego Inc Heterocycles n-substitues pour le traitement de l'hypercholesterolemie, de la dyslipidemie et autres troubles du metabolisme, du cancer et de pathologies diverses
GB0204252D0 (en) * 2002-02-23 2002-04-10 Oxford Glycosciences Uk Ltd Novel compounds
WO2003070238A1 (fr) 2002-02-23 2003-08-28 Oxford Glycosciences (Uk) Ltd Agents antifongiques a base de thiazolidine
US7102000B2 (en) * 2002-03-08 2006-09-05 Incyte San Diego Inc. Heterocyclic amide derivatives for the treatment of diabetes and other diseases
AU2002953533A0 (en) 2002-12-24 2003-01-16 Arthron Limited Fc receptor modulating compounds and compositions
US6794401B2 (en) * 2003-01-17 2004-09-21 Bexel Pharmaceuticals, Inc. Amino acid phenoxy ethers
US7521465B2 (en) * 2003-01-17 2009-04-21 Bexel Pharmaceuticals, Inc. Diphenyl ether derivatives
US7781464B2 (en) * 2003-01-17 2010-08-24 Bexel Pharmaceuticals, Inc. Heterocyclic diphenyl ethers
US20100305174A1 (en) 2005-04-13 2010-12-02 Orchid Research Laboratories Limited Novel Heterocyclic Derivatives
JP4224046B2 (ja) 2005-08-10 2009-02-12 シプロ化成株式会社 光学記録媒体
JP2007045946A (ja) 2005-08-10 2007-02-22 Shipro Kasei Kaisha Ltd チアゾリジン誘導体を用いた有機色素
US7268364B2 (en) * 2005-09-23 2007-09-11 Aculon, Inc. Hybrid devices
CA2628725A1 (fr) * 2005-11-15 2007-05-31 Glycofi, Inc. Production de glycoproteines a o-glycosylation reduite
SG153872A1 (en) * 2006-03-08 2009-07-29 Nat Inst Immunology 2-thioxothiazolidin-4-one compounds and compositions as antimicrobial and antimalarial agents targeting enoyl-acp reductase of type ii fatty acid synthesis pathway and other cell growth pathways
JP5131666B2 (ja) * 2006-05-16 2013-01-30 独立行政法人産業技術総合研究所 タンパク質の高分泌生産方法
ES2614931T3 (es) * 2006-08-04 2017-06-02 Beth Israel Deaconess Medical Center Inhibidores de la piruvato cinasa y métodos de tratamiento de enfermedad
MX2010012626A (es) * 2008-05-20 2011-02-22 Merck Sharp & Dohme Produccion eficiente de proteinas heterologas utilizando inhibidores de manosil transferasa.

Also Published As

Publication number Publication date
WO2009143041A1 (fr) 2009-11-26
CA2721459A1 (fr) 2009-11-26
JP2011520968A (ja) 2011-07-21
MX2010012626A (es) 2011-02-22
US8765409B2 (en) 2014-07-01
EP2300446A1 (fr) 2011-03-30
ATE536347T1 (de) 2011-12-15
CN102036974B (zh) 2014-07-23
US20110076721A1 (en) 2011-03-31
AU2009249314A1 (en) 2009-11-26
JP2014055157A (ja) 2014-03-27
US8309325B2 (en) 2012-11-13
US20130316398A1 (en) 2013-11-28
JP5411929B2 (ja) 2014-02-12
CN102036974A (zh) 2011-04-27

Similar Documents

Publication Publication Date Title
AU2006316838B2 (en) Production of glycoproteins with reduced O-glycosylation
US20120135461A1 (en) Production of glycoproteins with reduced o-glycosylation comprising the use of an alpha-1,2-mannosidase
US8771989B2 (en) Vectors and yeast strains for protein production: Ca2+ ATPase overexpression
US8765409B2 (en) Efficient production of heterologous proteins using mannosyl transferase inhibitors
EP2771479B1 (fr) Procédés pour augmenter l'occupation des n-glycanes et réduire la production des n-glycanes hybrides dans des souches de pichia pastoris n'exprimant pas alg3
EP2771461B1 (fr) Souches hôtes eucaryotes inférieures génétiquement modifiées pour une expression protéique recombinante
US20140302556A1 (en) Controlling o-glycosylation in lower eukaryotes
US20140287463A1 (en) Engineered pichia strains with improved fermentation yield and n-glycosylation quality
AU2012203729B2 (en) Production of glycoproteins with reduced O-glycosylation
CN101257922A (zh) 主要包含Man7GlcNAc2、Man8GlcNAc2糖形式的免疫球蛋白

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20101220

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

DAX Request for extension of the european patent (deleted)
GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009004093

Country of ref document: DE

Effective date: 20120209

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: MARKS & CLERK (LUXEMBOURG) LLP

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120307

LTIE Lt: invalidation of european patent or patent extension

Effective date: 20111207

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120308

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

REG Reference to a national code

Ref country code: CH

Ref legal event code: PFA

Owner name: MERCK SHARP & DOHME CORP.

Free format text: SCHERING CORPORATION#2000 GALLOPING HILL ROAD#KENILWORTH, NJ 07033 (US) -TRANSFER TO- MERCK SHARP & DOHME CORP.#126 EAST LINCOLN AVENUE#RAHWAY, NEW JERSEY 07065 (US)

Ref country code: CH

Ref legal event code: PFUS

Owner name: SCHERING CORPORATION, US

Free format text: FORMER OWNER: MERCK SHARP DOHME CORP., US

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120307

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120407

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: SCHERING CORPORATION

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: MERCK SHARP & DOHME CORP.

REG Reference to a national code

Ref country code: GB

Ref legal event code: 732E

Free format text: REGISTERED BETWEEN 20120802 AND 20120808

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120409

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

REG Reference to a national code

Ref country code: NL

Ref legal event code: SD

Effective date: 20120828

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 536347

Country of ref document: AT

Kind code of ref document: T

Effective date: 20111207

REG Reference to a national code

Ref country code: NL

Ref legal event code: TD

Effective date: 20120924

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

26N No opposition filed

Effective date: 20120910

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009004093

Country of ref document: DE

Representative=s name: ABITZ & PARTNER, DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120531

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009004093

Country of ref document: DE

Effective date: 20120910

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009004093

Country of ref document: DE

Representative=s name: ABITZ & PARTNER, DE

Effective date: 20121213

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004093

Country of ref document: DE

Owner name: MERCK SHARP & DOHME CORP., US

Free format text: FORMER OWNER: MERCK SHARP & DOHME CORP., RAHWAY, US

Effective date: 20121213

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004093

Country of ref document: DE

Owner name: SCHERING CORPORATION, US

Free format text: FORMER OWNER: MERCK SHARP & DOHME CORP., RAHWAY, US

Effective date: 20121213

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004093

Country of ref document: DE

Owner name: MERCK SHARP & DOHME CORP., RAHWAY, US

Free format text: FORMER OWNER: MERCK SHARP & DOHME CORP., RAHWAY, N.J., US

Effective date: 20121213

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009004093

Country of ref document: DE

Representative=s name: ABITZ & PARTNER PATENTANWAELTE MBB, DE

Effective date: 20121213

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

REG Reference to a national code

Ref country code: FR

Ref legal event code: TP

Owner name: SCHERING CORPORATION, US

Effective date: 20130318

Ref country code: FR

Ref legal event code: CD

Owner name: SCHERING CORPORATION, US

Effective date: 20130319

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120518

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120318

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009004093

Country of ref document: DE

Representative=s name: ABITZ & PARTNER, DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009004093

Country of ref document: DE

Representative=s name: ABITZ & PARTNER, DE

Effective date: 20130514

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004093

Country of ref document: DE

Owner name: MERCK SHARP & DOHME CORP., US

Free format text: FORMER OWNER: SCHERING CORPORATION, RAHWAY, US

Effective date: 20130514

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004093

Country of ref document: DE

Owner name: MERCK SHARP & DOHME CORP., RAHWAY, US

Free format text: FORMER OWNER: SCHERING CORPORATION, RAHWAY, US

Effective date: 20130514

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009004093

Country of ref document: DE

Representative=s name: ABITZ & PARTNER PATENTANWAELTE MBB, DE

Effective date: 20130514

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20130514

Year of fee payment: 5

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20111207

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090518

REG Reference to a national code

Ref country code: NL

Ref legal event code: V1

Effective date: 20141201

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20141201

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20150428

Year of fee payment: 7

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160531

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160531

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004093

Country of ref document: DE

Owner name: MERCK SHARP & DOHME LLC, RAHWAY, US

Free format text: FORMER OWNER: MERCK SHARP & DOHME CORP., RAHWAY, N.J., US

REG Reference to a national code

Ref country code: GB

Ref legal event code: 732E

Free format text: REGISTERED BETWEEN 20220908 AND 20220914

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230412

Year of fee payment: 15

Ref country code: DE

Payment date: 20230412

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230412

Year of fee payment: 15